The pair typically consists of:
11418-2-AP: Targets a fusion protein antigen (Ag1986) with predicted cross-reactivity across species .
EPR14439(B): Binds to a linear epitope in human COX5B, validated for specificity in mitochondrial protein complexes .
Application | 11418-2-AP | EPR14439(B) |
---|---|---|
Western Blot | 1:500–1:2,000 | 1:5,000–1:100,000 |
IHC | 1:250–1:1,000 | 1:500 (paraffin-embedded tissue) |
IF/ICC | 1:200–1:800 | 1:1,000 (cell-based assays) |
Observed Molecular Weight: 14 kDa in WB for both antibodies .
Species | 11418-2-AP | EPR14439(B) |
---|---|---|
Human | ✔️ (HeLa, liver cancer) | ✔️ (HepG2, HeLa) |
Mouse | ✔️ (liver tissue) | ❌ (No data) |
Rat | ✔️ (liver tissue) | ❌ (No data) |
COX5B in Respiratory Supercomplexes:
Interaction with MAVS:
COX5B and Bcl-2:
COX5B (Cytochrome c oxidase subunit 5B) is a nuclear-coded polypeptide chain of cytochrome c oxidase, the terminal oxidase in mitochondrial electron transport. It functions as a critical component of Complex IV in the respiratory chain, which catalyzes the reduction of oxygen to water. The COX complex consists of multiple subunits that work together to promote efficient oxidative phosphorylation .
COX5B is particularly important because:
It plays a crucial role in the electron transport chain that drives ATP production
It has emerged as a potential biomarker in several cancer types
Recent research has identified non-canonical functions beyond its role in bioenergetics, including regulation of antiviral signaling
It responds to oxygen levels in cells, with its expression being regulated differently under aerobic versus hypoxic conditions
COX5B antibodies have been validated for multiple applications across various experimental contexts:
When designing experiments, researchers should consider that different antibody clones may perform optimally under different conditions and applications .
Proper validation of COX5B antibody specificity is critical for experimental reliability:
Positive controls: Use tissues/cells known to express COX5B at high levels, such as HeLa cells, L02 cells, liver tissues (human/mouse/rat)
Knockdown/knockout validation: Compare antibody signal between wild-type and COX5B knockdown/knockout cells; properly validated antibodies should show decreased or absent signal in knockdown/knockout samples
Molecular weight verification: Confirm that the detected protein band appears at the expected molecular weight of 14 kDa
Multiple antibody comparison: When possible, verify results with at least two different antibody clones targeting different epitopes of COX5B
Species cross-reactivity testing: If working across species, validate each antibody's reactivity in the specific species being studied, as reactivity can vary significantly
Based on published protocols and commercial recommendations, the following conditions optimize Western blot detection of COX5B:
Sample preparation:
Mitochondrial enrichment often improves signal-to-noise ratio due to COX5B's mitochondrial localization
Standard RIPA or NP-40 based lysis buffers are generally suitable
Include protease inhibitors to prevent degradation
Electrophoresis and transfer:
Use 12-15% SDS-PAGE gels due to COX5B's relatively small molecular weight (14 kDa)
PVDF membranes typically yield better results than nitrocellulose for small proteins
Semi-dry transfer systems work well for COX5B
Antibody incubation and detection:
Primary antibody dilutions typically range from 1:500 to 1:2000
Overnight incubation at 4°C often yields cleaner results than shorter room temperature incubations
For polyclonal antibodies, longer blocking times (1-2 hours) with 5% non-fat milk or BSA may reduce non-specific binding
Troubleshooting multiple bands:
If detecting multiple bands, consider:
Using more stringent washing conditions
Trying a different antibody clone
Performing a knockdown validation to confirm the specific band
Reliable quantification of COX5B protein requires careful consideration of several factors:
Western blot quantification:
Always normalize to appropriate loading controls (β-actin for whole cell lysates, VDAC or other mitochondrial proteins for mitochondrial fractions)
Use linear range detection methods; avoid oversaturated bands
Consider running standard curves with known quantities of recombinant COX5B for absolute quantification
Immunohistochemical quantification:
Flow cytometry:
Permeabilization is critical as COX5B is intracellular
Include isotype controls to establish background staining levels
Consider dual staining with mitochondrial markers to confirm specificity
Real-time considerations:
Protein levels should be validated against mRNA expression when possible
Consider that post-translational regulation may cause discrepancies between mRNA and protein levels
Proper storage and handling are essential for maintaining antibody performance:
Storage conditions:
Store antibodies at -20°C in small aliquots to avoid repeated freeze-thaw cycles
Most COX5B antibodies are stable for at least one year when properly stored
Some commercial antibodies are supplied with glycerol (typically 50%) and can be stored at -20°C without aliquoting
Working solutions:
Prepare fresh working dilutions on the day of experiment
Store diluted antibody at 4°C for short-term use (1-2 weeks maximum)
Include preservatives (0.02% sodium azide) in working solutions for longer storage
Handling precautions:
Avoid contamination by using clean pipette tips
Centrifuge antibody vials briefly before opening to collect liquid at the bottom
Some antibody preparations contain BSA (0.1%) which may interfere with certain applications
COX5B has been identified as a negative regulator of MAVS (mitochondrial antiviral signaling protein) activity, revealing a novel function beyond its canonical role in oxidative phosphorylation:
Mechanism of interaction:
COX5B physically interacts with MAVS at the mitochondria through the CARD domain of MAVS
This interaction was confirmed by co-immunoprecipitation experiments in HEK293 cells and immunostaining analyses showing co-localization
The interaction appears to be specific to COX5B, as COX5A (another component of the CcO complex) did not co-immunoprecipitate with MAVS
Functional consequences:
Overexpression of COX5B suppresses MAVS-induced activation of IFN-β, NF-κB, and ISRE promoters
COX5B knockdown enhances antiviral signaling and reduces viral titers in infected cells
Mitochondrial localization of COX5B is critical for this regulatory function, as demonstrated by experiments with transit peptide mutants
Coordination with autophagy:
COX5B appears to coordinate with the autophagy pathway to control MAVS aggregation
This interaction provides a mechanistic link between mitochondrial electron transport, ROS production, autophagy, and antiviral immunity
These findings highlight an important immunoregulatory role for COX5B beyond its classical function in energy metabolism, suggesting that COX5B antibodies may be valuable tools in studying the crosstalk between metabolism and immunity.
Recent research has uncovered important roles for COX5B in cancer biology, particularly in colorectal cancers (CRCs):
Clinical significance:
Cellular mechanisms:
COX5B promotes cell growth and attenuates anticancer drug susceptibility in CRC cells
These effects appear to be mediated through COX5B-dependent regulation of Claudin-2 (CLDN2) expression
Silencing of COX5B represses cell growth and enhances the susceptibility of CRC cells to anticancer drugs
Bioenergetic connections:
COX5B expression correlates with oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) in tumor tissues
This suggests that COX5B-mediated metabolic reprogramming may contribute to cancer cell survival and drug resistance
These findings highlight the potential of COX5B as both a prognostic biomarker and therapeutic target in colorectal cancer, emphasizing the importance of reliable antibodies for studying its expression and localization in clinical samples.
Proper validation of COX5B knockdown/knockout is essential for functional studies:
Protein-level validation:
Western blotting using validated COX5B antibodies is the gold standard for confirming protein reduction
Flow cytometry can provide quantitative measurement at the single-cell level
Immunofluorescence microscopy allows visualization of knockdown efficiency and subcellular localization changes
Functional validation:
Cytochrome c oxidase activity assays directly measure the functional consequence of COX5B reduction
Protocols using isolated mitochondria and commercially available kits provide quantifiable measurements
Comparison of enzymatic activity between control and knockdown/knockout samples confirms functional impact
Validation controls:
Include multiple siRNA sequences targeting different regions of COX5B to control for off-target effects
Smart pool siRNAs with validated sequences (e.g., CGACUGGGUUGGAGAGGGA, GAGCACCUGCACUAAAUUA, GGGACUGGACCCAUACAAU, GAGAAUAGUAGGCUGCAUC) have been effectively used in published studies
Non-targeting control siRNAs are essential negative controls
Phenotypic validation:
ATP production measurement can confirm the metabolic impact of COX5B reduction
Studies have shown reduced ATP levels in COX5B knockdown cells, consistent with its role in oxidative phosphorylation
Different COX5B antibody clones may exhibit significant variations in performance across applications:
Epitope considerations:
Antibodies targeting different epitopes may yield different results depending on protein conformation or post-translational modifications
N-terminal antibodies may be affected by the presence/absence of the mitochondrial transit peptide
C-terminal antibodies may be affected by protein-protein interactions
Validation approaches:
When transitioning between antibody clones, researchers should perform side-by-side comparisons
Knockout/knockdown validation is particularly important when switching antibodies
Cross-validation with orthogonal techniques (e.g., mass spectrometry) provides highest confidence
Several methodological approaches have been successfully employed to study COX5B protein interactions:
Co-immunoprecipitation (Co-IP):
Successfully used to demonstrate COX5B-MAVS interactions in HEK293 cells
Both overexpressed epitope-tagged proteins and endogenous proteins can be studied
Controls should include reverse Co-IP (immunoprecipitating with anti-MAVS and blotting for COX5B) and negative controls (e.g., COX5A)
Proximity ligation assays:
Provide in situ visualization of protein-protein interactions
Particularly useful for detecting transient or weak interactions in their native cellular context
Requires validated antibodies raised in different host species
Subcellular co-localization:
Immunofluorescence microscopy has confirmed COX5B-MAVS co-localization at mitochondria
Confocal or super-resolution microscopy provides higher resolution of mitochondrial structures
Both GFP-tagged COX5B and antibody staining approaches have been validated
Domain mapping:
Truncation mutants (e.g., MAVSΔCARD) can identify specific interaction domains
For COX5B, the mitochondrial transit peptide (31-residue N-terminal sequence) is critical for proper localization and function
COX5B antibodies provide valuable tools for assessing mitochondrial function in various disease contexts:
Cancer metabolism studies:
IHC analysis of COX5B expression in patient tumor samples correlates with clinical outcomes and metabolic parameters
Comparison of tumor/non-tumor expression ratios provides greater predictive value than absolute expression levels
Combined analysis with bioenergetic measurements (OCR, ECAR) links COX5B to metabolic reprogramming in tumors
Neurodegenerative disease models:
COX5B antibodies can detect alterations in mitochondrial respiratory complexes
Dual staining with other mitochondrial markers can reveal respiratory chain defects
Quantification of COX5B levels may serve as a biomarker for mitochondrial dysfunction
Viral infection studies:
COX5B antibodies help elucidate the interplay between mitochondrial function and antiviral responses
Changes in COX5B-MAVS interactions during viral infection can be monitored using co-IP and immunofluorescence
Correlation with viral titers provides functional relevance of these interactions
Methodological considerations:
Researchers face several challenges when comparing COX5B expression data:
Tissue-specific expression patterns:
COX5B expression varies significantly across tissues and cell types
Higher expression is typically observed in metabolically active tissues like liver
Normalization to tissue-specific reference genes is critical for accurate comparisons
Technical variables:
Different antibody clones may have varying sensitivities and specificities
Fixation methods can significantly impact epitope accessibility in IHC applications
Sample preparation methods (particularly for mitochondrial proteins) can affect detection efficiency
Biological complexity:
COX5B functions within a multi-subunit complex, and its expression may be coordinated with other complex components
Post-translational modifications may affect antibody recognition but have functional significance
Subcellular localization changes (e.g., during stress conditions) may complicate interpretation
Solution approaches:
Use multiple antibodies targeting different epitopes when possible
Include appropriate positive controls (e.g., tissues known to express high COX5B levels)
Consider analysis of COX5B within the context of other mitochondrial proteins to distinguish specific versus general mitochondrial effects
When comparing across studies, account for methodological differences in sample preparation and detection
Single-cell approaches offer powerful new insights into COX5B biology:
Single-cell protein analysis:
Flow cytometry with COX5B antibodies can reveal population heterogeneity in mitochondrial content
Imaging flow cytometry combines quantitative measurement with visualization of subcellular localization
Mass cytometry (CyTOF) allows simultaneous detection of COX5B with many other cellular markers
Spatial transcriptomics/proteomics:
Emerging methods combining in situ hybridization with proteomics can map COX5B expression patterns within tissues
These approaches may reveal previously unrecognized spatial regulation of mitochondrial function
Single-cell applications in cancer research:
Analysis of COX5B at the single-cell level may identify resistant cell populations within tumors
Combined with functional assays, this could elucidate how metabolic heterogeneity contributes to treatment resistance
Methodological considerations:
Antibody validation at the single-cell level requires additional controls
Fixation and permeabilization protocols need optimization for mitochondrial proteins
Multiplexed approaches require careful antibody panel design to avoid spectral overlap
The emerging understanding of COX5B's roles beyond energy metabolism suggests several therapeutic opportunities:
Cancer therapy approaches:
COX5B knockdown sensitizes colorectal cancer cells to anticancer drugs, suggesting potential for combination therapies
The COX5B-CLDN2 axis represents a novel targetable pathway in colorectal cancer
Patient stratification based on COX5B expression could identify those most likely to benefit from metabolism-targeting therapies
Antiviral applications:
Modulating COX5B-MAVS interactions could potentially enhance antiviral immunity
Understanding how COX5B coordinates with autophagy to regulate antiviral signaling may reveal new therapeutic targets
Temporal regulation of these pathways could provide a means to boost initial antiviral responses while preventing chronic inflammation
Small molecule discovery:
Development of small molecules that specifically disrupt COX5B protein interactions rather than its metabolic function
High-throughput screening assays using COX5B antibodies could identify compounds that alter its expression or localization
Pharmacodynamic biomarkers:
COX5B antibodies could serve as tools for monitoring treatment efficacy in approaches targeting mitochondrial metabolism
Changes in COX5B expression or post-translational modifications might predict response to therapy